Você está na página 1de 10

Lee et al.

J Nanobiotechnol (2015) 13:54


DOI 10.1186/s12951-015-0118-0

REVIEW Open Access

Organic–inorganic hybrid nanoflowers:


types, characteristics, and future prospects
Seung Woo Lee1, Seon Ah Cheon1, Moon Il Kim2 and Tae Jung Park1* 

Abstract 
Organic–inorganic hybrid nanoflowers, a newly developed class of flower-like hybrid nanoparticles, have received
much attention due to their simple synthesis, high efficiency, and enzyme stabilizing ability. This article covers, in
detail, the types, structural features, mechanism of formation, and bio-related applications of hybrid nanoflowers. The
five major types of hybrid nanoflowers are discussed, i.e., copper–protein, calcium–protein, and manganese–protein
hybrid nanoflowers, copper–DNA hybrid nanoflowers, and capsular hybrid nanoflowers. The structural features of
these nanoflowers, such as size, shape, and protein ratio generally determine their applications. Thus, the specific
characteristics of hybrid nanoflowers are summarized in this review. The interfacial mechanism of nanoflower for-
mation is examined in three steps: first, combination of metal ion and organic matter; second, formation of petals;
third, growth of nanoflowers. The explanations provided herein can be utilized in the development of innovative
approaches for the synthesis of hybrid nanoflowers for prospective development of a plethora of hybrid nanoflowers.
The future prospects of hybrid nanoflowers in the biotechnology industry, medicine, sensing, and catalysis are also
discussed.
Keywords:  Biosynthesis, Nanoflowers, Organic–inorganic hybrid

Background structures of nanoflowers, and it is a formidable task to


Since the inceptive studies of new nanomaterials in the apply these particles in the field of biochemistry.
early 2000s, various nanomaterials with captivating Enzymes are biological species with excellent activity
morphologies such as the core–shell structure [1], and and substrate specificity, but their use is limited by cer-
Janus particle [2] have been developed. Among these tain drawbacks such as high sensitivity to the surround-
new nanostructures, the topographical features of nano- ing environment, low reproducibility of experimental
flowers have piqued the interests of scientists because outcomes, and the requirement for complex purifica-
the higher surface-to-volume ratio in comparison with tion processes. To facilitate application in drug delivery
spherical nanoparticles is beneficial to enhancement in systems or detectors, novel nanomaterials called “nano-
the efficiency of surface reactions. Therefore, extensive biomaterials” have been developed. These are also known
studies focusing on the applications of the nanoflow- as “organic–inorganic hybrid nanomaterials;” the name
ers have been undertaken. Despite the increasing inter- indicates that all of the inorganic nanoparticle compo-
est in nanoflowers, synthesis of these species requires nents are bound to organic materials. Current methods
very harsh conditions such as toxic organic solvents, for the preparation of nano-biomaterials can be classified
high temperature, and high pressure. Thus, it is difficult into four categories in terms of immobilization [3–6],
to control their morphological features for tailoring the conjugation [7–9], crosslinking [10–13], and self-assem-
bly [14]. Nano-biomaterials have numerous prospective
applications in catalysis [15–18], biosensors [19–22], and
drug delivery [23–26].
*Correspondence: tjpark@cau.ac.kr
1
Proteins as enzymes have a strong affinity for metal
Department of Chemistry, Chung-Ang University, 84 Heukseok‑ro,
Dongjak‑gu, Seoul 06974, Republic of Korea
ions as cofactor and thus, the stability of proteins is gen-
Full list of author information is available at the end of the article erally enhanced during immobilization onto the metal

© 2015 Lee et al. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://
creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided
you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate
if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/
zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Lee et al. J Nanobiotechnol (2015) 13:54 Page 2 of 10

surface by the interaction such as charge affinity, covalent Types of nanoflowers


bond, and structural coupling [27, 28]. However, immobi- Hybrid nanoflowers can be classified on the basis of
lized proteins exhibit lower activity than the free enzyme, the structure of the particles and the type of metal and
primarily due to the loss of their activity by changing the organic material used. The classifications are summa-
orientation during the immobilization process and mass- rized in Table 1.
transfer limitations on solid supports [29–32]. Single-
protein nanoparticles and nanogels are examples of novel General nanoflowers using copper(II) ions and proteins
nano-biomaterials that have been used as catalysts with Since the initial development of hybrid nanoflowers con-
highly preserved activities [3, 10–13]. The activity of sisting of copper(II) ions and proteins, these species have
enzymes in these nanoparticles and nanogels could be as been intensively studied by focusing on their efficiency
much as ~60–90 % of that of the free enzymes. In one of and stability [35–39]. Because many studies on copper–
the few examples where the activity is better than that of protein hybrid nanoflowers have been investigated well
the free enzyme, organophosphorus hydrolase embedded than those of other hybrid nanoflowers, the synthesis
in mesoporous silica demonstrated an activity of ~200 % mechanism and applications of copper–protein hybrid
in solution compared with the free hydrolase [33]. In nanoflowers are relatively well understood.
the case of trypsin, which is a hydrolyzing enzyme that Ge et  al. [35] first serendipitously discovered the
can digest itself, its immobilization on a solid support hybrid nanoflower and confirmed that the copper ion
increased the catalytic efficiency by thousands of times and protein create a new type of particle via interaction.
compared with that of free trypsin [34]. However, prob- Subsequently, they synthesized four types of hybrid nano-
lems such as loss of enzyme properties are still encoun- flowers using α-lactalbumin, laccase, carbonic anhydrase,
tered in these complicated processes. and lipase, respectively. The synthesized hybrid nanoflow-
Recently, a new approach for facile and safe synthe- ers were used in the detection of phenols and oxidation
sis of hybrid nanomaterials was developed to overcome of catecholamines. Each efficiency of the nanoflowers was
the limitations of conventional methods [35]. Because it found to be the same or superior to (95–650 %) to those
is possible to fabricate a nanomaterial simply by adding
protein to metal ion solution, this synthetic method does
not require any toxic elements or extreme harsh condi- Table 1  Types of organic–inorganic hybrid nanoflowers
tions. Therefore, the organic substance involved in the Metal ion Organic material Target References
synthesis is subjected to less manipulation compared
1 Copper α-Lactalbumin – [35]
with other conventional methods to maintain the activity
Laccase Catecholamines,
of the immobilized enzyme. phenols
Carbonic anhy-
The flower-like hybrid nanomaterials generated by drase
this process are called “organic–inorganic hybrid nano- Lipase
flowers” or “hybrid nanoflowers”. Their synthesis mech-
2 Copper Laccase Phenol [36]
anism, physical properties, protein activity, stability,
3 Copper Glucose oxidase Glucose [37]
and reproducibility have been intensively studied, and and HRPa
thus far, these species have exhibited significantly bet- 4 Copper Trypsin HRPa, BSAb [38]
ter properties than the free enzymes. The feasibility of 5 Copper HRPa Hydrogen perox- [39]
in vivo application of nanoflowers in protein complexes, ide, phenol
medicines, and serological studies is also a topic of inves- 6 Calcium α-Amylase Cnp-G3c [44]
tigation. However, a new paradigm shift is required in 7 Calcium Chitosan Hydrogen per- [45]
application of hybrid nanoflowers from enzyme stability oxide
improvements and efficient drug delivery systems to new 8 Manganese Immunoglobulin G Ractopamine [46]
horizons such as cell imaging, biosensor, and medical Anti-ractopamine
ab
approaches.
Bovine serum
This review presents an overview of several synthetic albumin
strategies that have been suggested to diversify nano- 9 Copper DNA Specific cell [55]
flowers, including their structural features. Moreover, the 10 Copper Bovine liver Hydrogen per- [56]
synthesis mechanism is confirmed through analysis. On catalase oxide
the basis of experimental and theoretical data, we pro- a
  Horseradish peroxidase
pose future prospects for novel nanoflowers and their b
  Bovine serum albumin
further enhancement. c
 2-Chloro-4-nitrophenylmaltotrioside
Lee et al. J Nanobiotechnol (2015) 13:54 Page 3 of 10

Fig. 1  Application cycle of the membrane with incorporated laccase nanoflowers: fabrication, use, rinse, and reuse. Phenol and ortho-, meta-, and
para-substituted phenols react with 4-aminoantipyrine to form colored compounds, which can be easily detected. Reproduced with permission
from Ref. [36]: Copyright 2013 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim

of the conventional free enzyme solutions. The increased or result in reduced enzymatic activity, which limits the
efficiency is derived from the following interplay of fac- application of the hybrid materials. As an alternative,
tors: (i) the large surface area of the nanoflower which the discovery of this novel nanoflower offers a fast and
does not cause mass-transfer limitations; (ii) the coop- simple synthesis. The procedure used in this experiment
erative interaction of the entrapped enzyme molecules; (Fig. 2) is as follows: glucose oxidized by GOx generates
(iii) the mutual influence of the enzyme and the micro- H2O2, which is immediately catalyzed by HRP to oxi-
environment of the nanoflower that contains metal ions dize 3,3′,5,5′-tetramethylbenzidine (TMB), resulting in a
on each other (for example, Cu2+ ions in the nanoflowers color change from clear to blue. The conventional two-
may enhance the activity of laccase). step process generally does not lead to high efficiency
Furthermore, the researchers developed a simplified owing to the diffusion of H2O2 generated from glucose
phenol detector comprising a syringe filter contain- oxidation. However, in the hybrid nanoflower, loss of the
ing adsorbed hybrid nanoflowers of laccase [36] (Fig. 1). generated H2O2 can be reduced because GOx and HRP
Briefly, a mixture of aqueous phenol and 4-aminoantipy- are simultaneously present, resulting in more accurate
rine was injected into the nanoflower-coated filter using a detection.
syringe and kept in the filter chamber for 5 min. The high The trypsin hybrid nanoflower synthesized by Lin
activity of laccase entrapped in the nanoflowers enabled et  al. was used for enhanced protein analysis [38]. Mass
rapid oxidative interaction of phenol with 4-aminoanti- spectrometry (MS) used widely for proteomic analysis,
pyrine to generate antipyrine dyes. The final red solution
was pushed out of the syringe and collected for analysis
by naked-eye visualization or by using a UV/Vis spectro-
photometer. The detection rate of this method is faster
than that of pre-existing gas chromatography (GC) or
liquid chromatography (LC). This method also facilitated
the reuse of the filter for about 1 month through cleaning
due to the high stability of the enzyme in the particles.
Following this germinal research, Sun et  al. synthe-
sized multienzymatic hybrid nanoflowers using glucose
oxidase (GOx) and horse-radish peroxidase (HRP) [37],
demonstrating the possibility that two or more enzymes
could be included in a single hybrid nanoflower. Even to
date, most co-immobilizations of multi-enzyme are per-
formed via comparatively complicated processes such as
by covalent cross-linking [40], encapsulation [41], gene Fig. 2  The cascade enzymatic reaction of multi-enzyme co-embed-
ded hybrid nanoflower for glucose detection. Reproduced with
fusion [42], and post-translational enzyme conjugation
permission from Ref. [37]: Copyright 2014, Royal Society of Chemistry
[43]. Unfortunately, these techniques are time consuming
Lee et al. J Nanobiotechnol (2015) 13:54 Page 4 of 10

involves analysis of enzymatically digested peptides cre-


ated from the parent protein. Therefore, effective prote-
olysis is required as a key step for effective identification
of proteins. Generally, proteolysis is performed by using
free enzymes, and suffers from some limitations such as
long digestion time, autolysis, low stability towards envi-
ronmental changes, and difficult recovery of products.
Enzymes immobilized on solid supports have been used
to overcome these disadvantages. The trypsin hybrid
nanoflower was first used as an immobilized enzyme by
Lin et  al. to achieve high proteolytic performance, good
enzyme stability and reusability, and a short digestion
time [38]. Moreover, the activity of the enzyme compo-
nent of the hybrid nanoflower was comparable to that Fig. 3  a Allosteric effect of α-amylase: α-amylase changes from
achieved in various conventional protein analyses. The inactive form to active form by binding calcium ion to allosteric site
in inactive α-amylase and b α-amylase immobilized in nanocrystals.
favorable results indicate that the hybrid nanoflower is a Reproduced with permission from Ref. [44]: Copyright 2013, American
promising enzyme system for proteomic analysis. Chemical Society
A hybrid nanoflower employing copper ions and HRP
for visual detection of hydrogen peroxide and phenol was
also synthesized [39]. As above, the hybrid nanoflower
was used as a catalyst in the reaction of hydrogen per- in an inactive state as the functional site is inhibited.
oxide and phenol in the presence of 4-aminoantipyrine. In contrast, in the presence of calcium ions, the allos-
Changes in the solution color could be detected with teric sites of α-amylase are occupied by the calcium ion,
concentrations as low as 0.5 μM hydrogen peroxide and thus affecting the structure of the functional site. Dur-
1 μM phenol by the naked eye. The threshold concentra- ing the synthesis of the hybrid nanoflower using calcium
tion of hydrogen peroxide to prevent cellular damage is ions and α-amylase, the calcium ions activate and are
50 μM; thus, the limit of detection (LOD) obtained with strongly bound to α-amylase. Because the two elements
the use of the hybrid nanoflower meets the requirement are located close to each other during the formation of
for early clinical diagnosis. The LOD for phenol also sat- the hybrid nanoflower, α-amylase was active for a longer
isfies the requirements for application to the detection of period than the free enzyme, which is randomly and
water pollution. In contrast, the LODs for hydrogen per- briefly activated by calcium ions (Fig.  3b). Thus, Wang
oxide and phenol using free HRP were ~20 and ~10 μM, et al. showed that it is essential to understand the chemi-
respectively. The enzyme activity of the HRP hybrid cal interaction between the protein and nanomaterials to
nanoflower is enhanced by the large surface-to-volume improve the functionality of the protein in a given envi-
ratio that facilitates contact with reactants. Moreover, the ronment [44].
reaction time of the HRP hybrid nanoflower (~5 min) is A hybrid nanoflower employing calcium ions was also
faster than that of free HRP (~25 min). The above results presented in a recent study [45]. Unlike previous syn-
revealed that the hybrid nanoflower is more useful for the thesis methods using protein solution, chitosan (CS)
development of a highly sensitive colorimetric sensing and tripolyphosphate (TPP) were used to fabricate a gel-
platform. form composite via ionic bonding. Although this method
has different steps from that used by Hou et al. [44], the
General nanoflowers using calcium(II) ions and proteins mechanism of nanoflower formation is the same (Fig. 4).
Nanoflowers employing calcium ions have also been Calcium ions and TPP form calcium phosphate crystals,
investigated, even though most nanoflowers are synthe- and the nanoflower is obtained by the reaction of these
sized with the copper ion [44, 45]. A nanoflower employ- crystals and the CS-TPP gel complex. Almost any type
ing calcium phosphate crystals was synthesized by Wang of catalyst could be used in this experiment because the
et  al. by using the same method used for the synthesis catalyst combines with chitosan in the CS-TPP gel com-
of copper nanoflowers [44]. The flower-like morphol- plex by electrostatic interaction. However, because addi-
ogy was confirmed and a postulate of how the activity tion of the catalyst declined in the nucleation sites for
increases due to the enzyme immobilization procedure calcium phosphate formation, it should be noted that the
was presented. The α-amylase used in this experiment is maximum amount of catalyst for the formation of the
an enzyme that exhibits allosteric phenomena (Fig.  3a). nanoflower was 5.0  mg. This technique for nanoflower
In the absence of calcium ions, α-amylases are usually synthesis provides a new approach that facilitates the
Lee et al. J Nanobiotechnol (2015) 13:54 Page 5 of 10

Fig. 4  Schematic synthesis of chitosan–calcium ion hybrid nanoflower. Chitosan binds to pyrophosphate through ionotropic gelation and gener-
ates CS-TPP gel complex which reacts with calcium phosphate crystal to form hybrid nanoflower. Reproduced with permission from Ref. [45]:
Copyright 2014, American Chemical Society

generation of the nanoflower using a variety of organic hybrid nanoflowers are more sensitive [52–54]. This
substances. study shows that the hybrid nanoflower can be used as
electrochemical tools as well as catalyst or drug delivery
General nanoflowers using manganese(II) ions and proteins matter.
Manganese phosphate hybrid nanoflowers were syn-
thesized by Zhang et  al. to apply into a novel electro- General nanoflowers using copper(II) ion and DNA
chemical biosensor for detection of ractopamine, which Hu et  al. used DNA as the organic material instead of
can cause acute poisoning consumed by humans [46]. protein in hybrid nanoflowers [55]. Because DNA is
Although many analytical methods have been devel- highly soluble in aqueous medium and has a high content
oped for the detection of ractopamine, such as liquid of nitrogen atoms in its structure like protein, it could
chromatography–mass spectrometry (LC–MS) [47, be used in the synthesis of hybrid nanoflowers by bind-
48], gas chromatography–mass spectrometry (GC– ing metal ions. The DNA hybrid nanoflower morphology
MS) [49], high-performance liquid chromatography was combined with the fluorescence resonance energy
(HPLC) [50], and bioassay [51]. They have some limits transfer (FRET) phenomenon to obtain high-resolution
owing to high instrument cost and long-time analysis. images of cells or to use for traceable drug delivery sys-
To solve the problem, electrochemical methods were tems. In brief, the researchers created a site in the DNA
applied by using the phenolic hydroxyl group, which template for the simultaneous attachment of a drug and
makes ractopamine electrochemically active [52]. How- fluorescent dyes (FAM, CY3, ROX), and subsequently,
ever, this detection method was also limited that due to they synthesized a hybrid nanoflower coupled to the
its low response activity on electrode surfaces. Hybrid fabricated DNA by rolling circle replication (RCR) with
nanoflower as a novel electrochemical biosensor over- the metal ions (Fig.  6). Consequently, they were able to
comes the disadvantage. These nanoflowers are made by obtain a high-resolution image based on FRET between
manganese(II) ions and three types of protein, immu- the dyes using long-wavelength light, which does not
noglobulin G (IgG), ractopamine antibody (RACanti), affect the cells. Furthermore, the path of drug delivery in
and bovine serum albumin (BSA) (Fig.  5). The detec- the living cells was successfully traced by monitoring the
tion limits of the developed electrochemical biosensors light emitted by the dyes. Thus, Hu et al. suggested that
were 4.6, 9.32, and 26 pg mL−1, respectively. Compared DNA hybrid nanoflowers can be applied in many fields
with the detection limits of previous different methods, by showing the feasibility of the synthesis of DNA hybrid
these results present that electrochemical methods using nanoflowers [55].
Lee et al. J Nanobiotechnol (2015) 13:54 Page 6 of 10

Fig. 5  Schematic representation of the synthesis of manganese-based hybrid nanoflowers as a novel electrochemical biosensor for the detection
of ractopamine, including (i) Mn3(PO4)2-IgG, (ii) Mn3(PO4)2-RACanti (anti-ractopamine antibody), and (iii) Mn3(PO4)2-BSA-Au nanoflowers. Reproduced
with permission from Ref. [46]: Copyright 2015, Elsevier

Fig. 6  Sequence-independent self-assembly of multicolor FRET (fluorescence resonance energy transfer) DNA hybrid nanoflower. Reproduced with
permission from Ref. [55]: Copyright 2014, Wiley-VCH Verlag

Capsular nanoflowers using copper(II) ions and proteins bio-chemical applications such as catalysis and drug
A capsular nanoflower was synthesized by Jiang and co- delivery.
workers, as shown in Fig. 7 [56, 57]. Compared with early
hybrid nanoflowers synthesized from a metal phosphate Characteristics
and protein, the capsular nanoflower was synthesized by Structural features and enzyme efficiency
an additional wrapping with protamine and silica through As shown in Table 2, the hybrid nanoflowers can be clas-
a biomimetic mineralization approach and removing the sified based on characteristics such as shape, size, pro-
metal from the core. Because the rough surface of the mul- tein/total weight ratio percent, and enzyme efficiency
tiple shells facilitates the adsorption of substrates, the cap- compared to that of the free enzyme.
sular nanoflower exhibited significantly enhanced enzyme Although the size of hybrid nanoflowers varies depend-
activity as well as better stability under extreme conditions ing on the protein concentration, the range of variation is
(high temperature, pH, long-term storage) than simple narrow (about ±3 μm). Thus, based on the average size
hybrid nanoflowers. These properties provide a practi- from optimized data, the nanoflowers range from 2 to
cable and useful solution for enhancing the efficiency in 30  μm, and the pore size (which is the diameter of the
Lee et al. J Nanobiotechnol (2015) 13:54 Page 7 of 10

Fig. 7  Scheme of preparation procedure of the FPSH capsules: a formation of protein–inorganic hybrid microflowers; b formation of (protamine-
silica)2 bilayers on the microflowers; c formation of the FPSH capsules after eliminating the microflower template through EDTA treatment. Repro-
duced with permission from Ref. [56]: Copyright 2014, The Royal Society of Chemistry

hole between petals) is approximately 0.1  μm. Because Mechanism


the size of most of these hybrid flowers is expressed in The mechanism for the synthesis of organic–inorganic
micro units (μm), we believe that the use of the term hybrid nanoflowers comprises three steps (Fig.  8). In
“microflower” is more correct than “nanoflower.” In addi- the early growth step, primary crystals of metal phos-
tion, the shape of the hybrid nanoflowers is similar to phate [M3(PO4)2, (M: Cu, Ca)] are formed. At this stage,
that of round flowers, but with subtle variations. Thus, the organic molecules (protein, DNA) form complexes
the pictures that resembles the respective scanning elec- with the metal ions (Cu2+, Ca2+), mainly through coor-
tron microscopy (SEM) images are provided to highlight dination via the amide groups in the protein backbone.
the differences (Table 2). These complexes provide a location for nucleation of the
The weight ratio of protein to total particle weight primary crystals. In the second growth stage, metal-pro-
measured by thermogravimetric analysis (TGA) is also tein crystals aggregate into large agglomerates of protein
summarized in Table  2, demonstrating that the protein molecules and primary petals are formed. The kinetically
constitutes 10–66 % of the total weight. Furthermore, as controlled growth of metal phosphate crystals originates
more protein is used, the percentage of protein increases. on the surfaces that have the Cu2+ binding sites of these
However, the encapsulation efficiency (the ratio of the agglomerates, causing flower-like petals to appear in the
amount of immobilized protein to the total amount of embryo. In the final step, anisotropic growth leads to
protein employed) follows a completely opposite trend. complete formation of a branched flower-like structure.
Excessive addition of protein to a constant weight of In this growth process, the protein induces nucleation of
inorganic component induces a dramatic decrease of the the metal phosphate crystals to form the scaffold for the
encapsulation efficiency. In light of these considerations, petals and serves as a “glue” to bind the petals together. In
the proper amount of protein must be selected to con- the absence of the proteins, large crystals, but no nano-
serve the protein and to attain good morphology and a flowers, are formed.
high weight percentage.
Finally, we consider the enzyme efficiency of hybrid Future prospects
nanoflowers. Compared with the corresponding free In the 1960s, the development of enzyme immobilization
enzyme solutions, the enzyme efficiency of hybrid nano- technology inspired chemists to design new biomaterials
flowers varies from 85 to over 1000 %. These results sug- containing highly stabilized enzymes. However, immo-
gest that the proteins in hybrid nanoflowers have higher bilization generally led to loss of the enzyme activity. By
activity and stability than the corresponding free enzyme the 1990s, the activity of enzymes immobilized on nano-
solutions despite immobilization in the flower petals. sized materials could be maintained at levels up to that
Moreover, hybrid nanoflowers overcome the previously of the free enzymes. Interest in this field was again awak-
encountered problem of mass-transfer limitation and ened with the unearthing of the first organic–inorganic
open up an avenue for application to various research hybrid nanoflower in 2012, and many studies focusing on
and detection fields. new flower-like hybrid nanomaterials are still in progress.
Lee et al. J Nanobiotechnol (2015) 13:54 Page 8 of 10

Table 2  Characteristics of organic–inorganic hybrid nanoflowers


Size Refer
Shape SEM image Protein ratio Enzyme efficiency
(μm) ences
Laccase: 650%
Carbonic anhydrase:
1 3 – [35]
260%
Lipase: 95%

2 20 – GOx & HRP: 400% [37]

3 27 3 9~12.6% Trypsin: 270% [38]

4 17 17.2% HRP: 506% [39]

5 4 0.5 20.16% α-Amylase: 1000% [44]

6 4 1 25.6% Catalase: 85% [45]

7 2 – – [55]

FPSH :
8 10 27.2% Catalase: 11% [56]
PSH : 66.5%
a
 Rose
b
  Blue ball
c
 Peony
d
 Satin
e
  Water flower
f
 Zonal
g
  Viburnum opulus
h
 Marigolds

Future research for the development of drug delivery biocompatibility, and modifications of hybrid nanoflower
systems, biosensors, biocatalysts, and bio-related devices structures and properties, should receive increasing
is anticipated to take multiple directions. New synthesis attention.
principles, new types of hybrid nanoflowers, and detailed
mechanisms are expected to emerge. The application of Conclusions
nanoflowers in bio-catalysis and enzyme mimetics, tis- In summary, organic–inorganic hybrid nanoflowers
sue engineering, and the design of highly sensitive bio- have piqued the interest of researches and numerous
sensing kits, as well as industrial bio-related devices with related papers have been published. Research in this
advanced functions, various and controllable syntheses, field is spurred by the simplicity of the synthesis and safe
Lee et al. J Nanobiotechnol (2015) 13:54 Page 9 of 10

Fig. 8  Synthesis mechanism of organic–inorganic hybrid nanoflower. Reproduced with permission from Ref. [35]: Copyright 2012, Nature Publish-
ing Group

conditions. Moreover, high efficiency and enzyme sta- 5. Asuri P, Karajanagi SS, Dordick JS, Kane RS. Directed assembly of carbon
nanotubes at liquid–liquid interfaces: nanoscale conveyors for interfacial
bility are readily achieved with hybrid nanoflowers. We biocatalysis. J Am Chem Soc. 2006;128(4):1046–7.
believe that the study of organic–inorganic hybrid nano- 6. Ji PJ, Tan HS, Xu X, Feng W. Lipase covalently attached to multiwalled
flowers will lead to creative solutions and rapid develop- carbon nanotubes as an efficient catalyst in organic solvent. AIChE J.
2010;56(11):3005–11.
ment of biomaterials and biotechnology industries. 7. Abuchowski A, McCoy JR, Palczuk NC, van Es T, Davis FF. Effect of covalent
attachment of polyethylene glycol on immunogenicity and circulating
life of bovine liver catalase. J Biol Chem. 1977;252(11):3582–6.
Abbreviations 8. Ehrat M, Luisi PL. Synthesis and spectroscopic characterization of insulin
GOx: glucose oxidase; HRP: horse-radish peroxidase; LOD: limit of detection; derivatives containing one or 2 poly(ethylene oxide) chains at specific
CS: chitosan; TPP: tripolyphosphate; SEM: scanning electron microscopy. positions. Biopolymers. 1983;22(1):569–73.
9. Boyer C, Bulmus V, Liu J, Davis TP, Stenzel MH, Barner-Kowollik C. Well-
Authors’ contributions defined protein–polymer conjugates via in situ RAFT polymerization. J
SWL and TJP conceived the idea of the work and performed the check- Am Chem Soc. 2007;129(22):7145–54.
ing the cited references. SAC and MIK were reviewed and commented the 10. Yan M, Ge J, Liu Z, Ouyang P. Encapsulation of single enzyme in nanogel
manuscript. All authors contributed to the preparation of the manuscript with enhanced biocatalytic activity and stability. J Am Chem Soc.
and commented on the final version. All authors read and approved the final 2006;128(34):11008–9.
manuscript. 11. Ge J, Lu D, Wang J, Yan M, Lu Y, Liu Z. Molecular fundamentals of enzyme
nanogels. J Phys Chem B. 2008;112(45):14319–24.
Author details 12. Ge J, Lu D, Wang J, Liu Z. Lipase nanogel catalyzed transesterification in
1
 Department of Chemistry, Chung-Ang University, 84 Heukseok‑ro, anhydrous dimethyl sulfoxide. Biomacromolecules. 2009;10(6):1612–8.
Dongjak‑gu, Seoul 06974, Republic of Korea. 2 Department of BioNano Tech- 13. Yan M, Liu Z, Lu D, Liu Z. Fabrication of single carbonic anhydrase
nology, Gachon University, 1342 Seongnamdaero, Sujeong‑gu, Seongnam‑si, nanogel against denaturation and aggregation at high temperature.
Gyeonggi‑do 461‑701, Republic of Korea. Biomacromolecules. 2007;8(2):560–5.
14. Shen L, Bao N, Prevelige PE, Gupta A. Fabrication of ordered nanostruc-
Acknowledgements tures of sulfide nanocrystal assemblies over self-assembled genetically
This research was supported by the Advanced Production Technology Devel- engineered P22 coat protein. J Am Chem Soc. 2010;132(49):17354–7.
opment Program, Ministry of Agriculture, Food and Rural Affairs (312066-3), 15. Kouassi GK, Irudayaraj J, McCarty G. Examination of Cholesterol
a grant of the Korean Health Technology R&D Project, Ministry of Health & oxidase attachment to magnetic nanoparticles. J Nanobiotechnology.
Welfare, Republic of Korea (HI13C0862) and the Chung-Ang University Gradu- 2005;3(1):1.
ate Research Scholarship in 2015. 16. Prakasham RS, Devi GS, Rao CS, Sivakumar VS, Sathish T, Sarma PN. Nickel-
impregnated silica nanoparticle synthesis and their evaluation for bio-
Compliance with ethical guidelines catalyst immobilization. Appl Biochem Biotechnol. 2010;160(7):1888–95.
17. Ding H, Wen L, Chen J. Porous silica nano-tube as host for enzyme immo-
Competing interests bilization. China Particuol. 2004;2(6):270–3.
The authors declare that they have no competing interests. 18. Ansari SA, Husain Q. Potential applications of enzymes immobilized on/in
nano materials: a review. Biotechnol Adv. 2012;30(3):512–23.
Received: 5 August 2015 Accepted: 25 August 2015 19. Wang R, Tian Z, Chen L. Nano-encapsulations liberated from barley pro-
tein microparticles for oral delivery of bioactive compounds. Int J Pharm.
2011;406(1–2):153–62.
20. Kumar PS, Ramakrishna S, Saini TR, Diwan PV. Influence of microencap-
sulation method and peptide loading on formulation of poly(lactide-co-
References glycolide) insulin nanoparticles. Pharmazie. 2006;61(7):613–7.
1. Schärtl W. Crosslinked spherical nanoparticles with core-shell topology. 21. Wanakule P, Liu GW, Fleury AT, Roy K. Nano-inside-micro: disease-respon-
Adv Mater. 2000;12(24):1899–908. sive microgels with encapsulated nanoparticles for intracellular drug
2. Lattuada M, Hatton TA. Synthesis, properties and applications of Janus delivery to the deep lung. J Control Release. 2012;162(2):429–37.
nanoparticles. Nano Today. 2011;6(3):286–308. 22. Kim JK, Anderson J, Jun HW, Repka MA, Jo S. Self-assembling peptide
3. Kim J, Grate JW. Single-enzyme nanoparticles armored by a nanometer- amphiphile-based nanofiber gel for bioresponsive cisplatin delivery. Mol
scale organic/inorganic network. Nano Lett. 2003;3(9):1219–22. Pharm. 2009;6(3):978–85.
4. Dyal A, Loos K, Noto M, Chang SW, Spagnoli C, Shafi KV, et al. Activity of 23. Njagi J, Andreescu S. Stable enzyme biosensors based on chemically
Candida rugosa lipase immobilized on γ-Fe2O3 magnetic nanoparticles. J synthesized Au-polypyrrole nanocomposites. Biosens Bioelectron.
Am Chem Soc. 2003;125(7):1684–5. 2007;23(2):168–75.
Lee et al. J Nanobiotechnol (2015) 13:54 Page 10 of 10

24. Lin J, Qu W, Zhang S. Disposable biosensor based on enzyme immo- 43. Hirakawa H, Kamiya N, Tanaka T, Nagamune T. Intramolecular electron
bilized on Au-chitosan-modified indium tin oxide electrode with flow transfer in a cytochrome P450cam system with a site-specific branched
injection amperometric analysis. Anal Biochem. 2007;360(2):288–93. structure. Protein Eng Des Sel. 2007;20(9):453–9.
25. Zhang YW, Zhang Y, Wang H, Yan B, Shen GL, Yu RQ. An enzyme immobi- 44. Wang LB, Wang YC, He R, Zhuang A, Wang X, Zeng J, et al. A new nano-
lization platform for biosensor designs of direct electrochemistry using biocatalytic system based on allosteric effect with dramatically enhanced
flower-like ZnO crystals and nano-sized gold particles. J Electroanal enzymatic performance. J Am Chem Soc. 2013;135(4):1272–5.
Chem. 2009;627(1–2):9–14. 45. Wang X, Shi J, Li Z, Zhang S, Wu H, Jiang Z, et al. Facile one-pot prepara-
26. Takhistov P. Electrochemical synthesis and impedance characteriza- tion of chitosan/calcium pyrophosphate hybrid microflowers. ACS Appl
tion of nano-patterned biosensor substrate. Biosens Bioelectron. Mater Interf. 2014;6(16):14522–32.
2004;19(11):1445–56. 46. Zhang Z, Zhang Y, Song R, Wang M, Yan F, He L, et al. Manganese(II) phos-
27. Sassolas A, Blum LJ, Leca-Bouvier BD. Immobilization strategies to phate nanoflowers as electrochemical biosensors for the high-sensitivity
develop enzymatic biosensors. Biotechnol Adv. 2012;30(3):489–511. detection of ractopamine. Sens Actuat B Chem. 2015;211:310–7.
28. Datta S, Christena LR, Rajaram YRS. Enzyme immobilization: an overview 47. Blanca J, Munoz P, Morgado M, Mendez N, Aranda A, Reuvers T, et al.
on techniques and support materials. Biotech. 2012;3(1):1–9. Determination of clenbuterol, ractopamine and zilpaterol in liver and
29. Kim J, Grate JW, Wang P. Nanobiocatalysis and its potential applications. urine by liquid chromatography tandem mass spectrometry. Anal Chim
Trend Biotechnol. 2008;26(11):639–46. Acta. 2005;529(1–2):199–205.
30. Ge J, Lu DN, Liu ZX, Liu Z. Recent advances in nanostructured biocata- 48. Shishani E, Chai SC, Jamokha S, Aznar G, Hoffman MK. Determination of
lysts. Biochem Eng J. 2009;44(1):53–9. ractopamine in animal tissues by liquid chromatography-fluorescence
31. Luckarift HR, Spain JC, Naik RR, Stone MO. Enzyme immobilization in a and liquid chromatography/tandem mass spectrometry. Anal Chim Acta.
biomimetic silica support. Nat Biotechnol. 2004;22(2):211–3. 2003;483(1–2):137–45.
32. Mateo C, Grazu V, Palomo JM, Lopez-Gallego F, Fernandez-Lafuente R, 49. He L, Su Y, Zeng Z, Liu Y, Huang X. Determination of ractopamine and
Guisan JM. Immobilization of enzymes on heterofunctional epoxy sup- clenbuterol in feeds by gas chromatography–mass spectrometry. Anim
ports. Nat Protoc. 2007;2(5):1022–33. Feed Sci Technol. 2007;132(3–4):316–23.
33. Lei C, Shin Y, Liu J, Ackerman EJ. Entrapping enzyme in a functionalized 50. Shelver WL, Smith DJ. Determination of ractopamine in cattle and sheep
nanoporous support. J Am Chem Soc. 2002;124(38):11242–3. urine samples using an optical biosensor analysis: comparative study
34. Dulay MT, Baca QJ, Zare RN. Enhanced proteolytic activity of cova- with HPLC and ELISA. J Agric Food Chem. 2003;51(13):3715–21.
lently bound enzymes in photopolymerized sol gel. Anal Chem. 51. Liu M, Ning BA, Qu LJ, Peng Y, Dong JW, Gao N, et al. Development of
2005;77(14):4604–10. indirect competitive immunoassay for highly sensitive determination of
35. Ge J, Lei J, Zare RN. Protein–inorganic hybrid nanoflowers. Nat Nanotech- ractopamine in pork liver samples based on surface plasmon resonance
nol. 2012;7(7):428–32. sensor. Sens Actuat B Chem. 2012;161(1):124–30.
36. Zhu L, Gong L, Zhang Y, Wang R, Ge J, Liu Z, Zare RN. Rapid detection of 52. Wu C, Sun D, Li Q, Wu KB. Electrochemical sensor for toxic ractopamine
phenol using a membrane containing laccase nanoflowers. Chem Asian and clenbuterol based on the enhancement effect of graphene oxide.
J. 2013;8(10):2358–60. Sens Actuat B Chem. 2012;168:178–84.
37. Sun J, Ge J, Liu W, Lan M, Zhang H, Wang P, et al. Multi-enzyme co- 53. Duan JH, He DW, Wang WS, Liu YC, Wu HP, Wang YS, et al. Glassy carbon
embedded organic–inorganic hybrid nanoflowers: synthesis and applica- electrode modified with gold nanoparticles for ractopamine and
tion as a colorimetric sensor. Nanoscale. 2014;6(1):255–62. metaproterenol sensing. Chem Phys Lett. 2013;574:83–8.
38. Lin Z, Xiao Y, Wang L, Yin Y, Zheng J, Yang H, et al. Facile synthesis of 54. Jin W, Yang G, Shao H, Qin A. A label-free impedimetric immunosensor for
enzyme-inorganic hybrid nanoflowers and their application as an immo- detection of 1-aminohydantoin residue in food samples based on sol–gel
bilized trypsin reactor for highly efficient protein digestion. RSC Adv. embedding antibody. Food Control. 2014;39:185–91.
2014;4(27):13888–91. 55. Hu R, Zhang X, Zhao Z, Zhu G, Chen T, Fu T, et al. DNA nanoflowers for
39. Lin Z, Xiao Y, Yin Y, Hu W, Liu W, Yang H. Facile synthesis of enzyme–inor- multiplexed cellular imaging and traceable targeted drug delivery.
ganic hybrid nanoflowers and its application as a colorimetric platform Angew Chem Int Ed. 2014;53(23):5821–6.
for visual detection of hydrogen peroxide and phenol. ACS Appl Mater 56. Shi JF, Zhang SH, Wang XL, Yang C, Jiang ZY. Preparation and enzymatic
Interf. 2014;6(13):10775–82. application of flower-like hybrid microcapsules through a biomimetic
40. Manesh KM, Santhosh P, Uthayakumar S, Gopalan AI, Lee KP. One-pot mineralization approach. J Mater Chem B. 2014;2(27):4289–96.
construction of mediatorless bi-enzymatic glucose biosensor based on 57. Li J, Jiang ZY, Wu H, Zhang L, Long LH, Jiang YJ. Constructing inorganic
organic–inorganic hybrid. Biosens Bioelectron. 2010;25(7):1579–86. shell onto LBL microcapsule through biomimetic mineralization: a
41. Delaittre G, Reynhout IC, Cornelissen JJ, Nolte RJ. Cascade reactions in an novel and facile method for fabrication of microbioreactors. Soft Matter.
all-enzyme nanoreactor. Chem Eur J. 2009;15(46):12600–3. 2010;6(3):542–50.
42. Fan Z, Wagschal K, Chen W, Montross MD, Lee CC, Yuan L. Multimeric
hemicellulases facilitate biomass conversion. Appl Environ Microbiol.
2009;75(6):1754–7.

Submit your next manuscript to BioMed Central


and take full advantage of:

• Convenient online submission


• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution

Submit your manuscript at


www.biomedcentral.com/submit

Você também pode gostar